We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Short CommunicationOpen Accesscc iconby iconnc iconnd icon

Novel hydrogel system eliminates subculturing and improves retention of nonsenescent mesenchymal stem cell populations

    Jacob G Hodge

    Bioengineering Graduate Program, University of Kansas, Lawrence, KS 66045, USA

    Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA

    ,
    Jennifer L Robinson

    Bioengineering Graduate Program, University of Kansas, Lawrence, KS 66045, USA

    Department of Chemical & Petroleum Engineering, University of Kansas, Lawrence, KS 66045, USA

    &
    Adam J Mellott

    *Author for correspondence: Tel.: +1 913 588 8308;

    E-mail Address: amellott@kumc.edu

    Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA

    Ronawk, LLC, Olathe, KS 66062, USA

    Published Online:https://doi.org/10.2217/rme-2022-0140

    Aim: To compare the physiological behavior of mesenchymal stem/stromal cells (MSCs) within an expandable tissue-mimetic 3D system relative to in vitro expansion in a traditional 2D system. Methods: Adipose-derived MSCs (ASCs) were continuously cultured for 6 weeks on either 2D culture plastic or in a 3D hydrogel system that eliminated subculturing. ASCs were assessed for senescence, ‘stem-like’ MSC markers, and ability for their secretome to augment a secondary cell population. Results: The 3D hydrogel system resulted in an enhanced retention of more regenerative, nonsenescent ASC populations that exhibited increased expression of ‘stem-like’ MSC surface markers. Conclusion: This study introduces a proof-of-concept design for a novel modular 3D system that can improve in vitro expansion of stem-like cell populations for future regenerative therapies.

    Tweetable abstract

    Novel tissue-mimetic 3D hydrogel system enhances the retention of nonsenescent ASC populations in vitro for up to 6 weeks in culture and eliminates the need to subculture, improving regenerative capacity of ASCs and their secreted biologics.

    Papers of special note have been highlighted as: • of interest

    References

    • 1. Hass R, Kasper C, Bohm S, Jacobs R. Different populations and sources of human mesenchymal stem cells (MSC): a comparison of adult and neonatal tissue-derived MSC. Cell Commun. Signal. 9, 12 (2011). • Review of mesenchymal stem/stromal cells (MSCs) and the different types of MSC populations, which also discusses the current and future role of MSC-derived regenerative therapies.
    • 2. Han Y, Li X, Zhang Y, Han Y, Chang F, Ding J. Mesenchymal stem cells for regenerative medicine. Cells 8(8), 886 (2019).
    • 3. Pittenger MF, Discher DE, Peault BM, Phinney DG, Hare JM, Caplan AI. Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen. Med. 4, 22 (2019).
    • 4. Musial-Wysocka A, Kot M, Majka M. The pros and cons of mesenchymal stem cell-based therapies. Cell Transplant. 28(7), 801–812 (2019).
    • 5. Lukomska B, Stanaszek L, Zuba-Surma E, Legosz P, Sarzynska S, Drela K. Challenges and controversies in human mesenchymal stem cell therapy. Stem Cells Int. 2019, 9628536 (2019).
    • 6. Koh J, Griffin DR, Archang MM et al. Enhanced in vivo delivery of stem cells using microporous annealed particle scaffolds. Small 15(39), e1903147 (2019).
    • 7. Choe G, Park J, Park H, Lee JY. Hydrogel biomaterials for stem cell microencapsulation. Polymers (Basel) 10(9), 997 (2018).
    • 8. Sivaraj D, Chen K, Chattopadhyay A et al. Hydrogel scaffolds to deliver cell therapies for wound healing. Front. Bioeng. Biotechnol. 9, 660145 (2021).
    • 9. Trounson A, McDonald C. Stem cell therapies in clinical trials: progress and challenges. Cell Stem Cell 17(1), 11–22 (2015).
    • 10. Walter MN, Wright KT, Fuller HR, Macneil S, Johnson WE. Mesenchymal stem cell-conditioned medium accelerates skin wound healing: an in vitro study of fibroblast and keratinocyte scratch assays. Exp. Cell Res. 316(7), 1271–1281 (2010).
    • 11. Watson SL, Marcal H, Sarris M, Di Girolamo N, Coroneo MT, Wakefield D. The effect of mesenchymal stem cell conditioned media on corneal stromal fibroblast wound healing activities. Br. J. Ophthalmol. 94(8), 1067–1073 (2010).
    • 12. Hu L, Wang J, Zhou X et al. Exosomes derived from human adipose mensenchymal stem cells accelerates cutaneous wound healing via optimizing the characteristics of fibroblasts. Sci. Rep. 6, 32993 (2016).
    • 13. Eleuteri S, Fierabracci A. Insights into the secretome of mesenchymal stem cells and its potential applications. Int. J. Mol. Sci. 20(18), 4597 (2019).
    • 14. Kabat M, Bobkov I, Kumar S, Grumet M. Trends in mesenchymal stem cell clinical trials 2004–2018: is efficacy optimal in a narrow dose range? Stem Cells Transl. Med. 9(1), 17–27 (2020).
    • 15. Ahmed AS, Sheng MH, Wasnik S, Baylink DJ, Lau KW. Effect of aging on stem cells. World J. Exp. Med. 7(1), 1–10 (2017).
    • 16. Jiang T, Xu G, Wang Q et al. In vitro expansion impaired the stemness of early passage mesenchymal stem cells for treatment of cartilage defects. Cell Death Dis. 8(6), e2851 (2017).
    • 17. Yin Q, Xu N, Xu D et al. Comparison of senescence-related changes between three- and two-dimensional cultured adipose-derived mesenchymal stem cells. Stem Cell Res. Ther. 11(1), 226 (2020).
    • 18. Drela K, Stanaszek L, Nowakowski A, Kuczynska Z, Lukomska B. Experimental strategies of mesenchymal stem cell propagation: adverse events and potential risk of functional changes. Stem Cells Int. 2019, 7012692 (2019). • Overview of current and ongoing limitations with MSC-derived therapies, with one major limitation being the non-standardized ex vivo expansion of MSCs that is needed for production of large-scale cell numbers and the adverse effects associated with this process.
    • 19. Izadpanah R, Kaushal D, Kriedt C et al. Long-term in vitro expansion alters the biology of adult mesenchymal stem cells. Cancer Res. 68(11), 4229–4238 (2008).
    • 20. Baxter MA, Wynn RF, Jowitt SN, Wraith JE, Fairbairn LJ, Bellantuono I. Study of telomere length reveals rapid aging of human marrow stromal cells following in vitro expansion. Stem Cells 22(5), 675–682 (2004).
    • 21. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell 126(4), 677–689 (2006).
    • 22. Dupont S. Role of YAP/TAZ in cell–matrix adhesion-mediated signalling and mechanotransduction. Exp. Cell Res. 343(1), 42–53 (2016).
    • 23. Vining KH, Mooney DJ. Mechanical forces direct stem cell behaviour in development and regeneration. Nat. Rev. Mol. Cell Biol. 18(12), 728–742 (2017).
    • 24. Ryu NE, Lee SH, Park H. Spheroid culture system methods and applications for mesenchymal stem cells. Cells 8(12), 1620 (2019).
    • 25. Edmondson R, Broglie JJ, Adcock AF, Yang L. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev. Technol. 12(4), 207–218 (2014).
    • 26. Zhou Y, Chen H, Li H, Wu Y. 3D culture increases pluripotent gene expression in mesenchymal stem cells through relaxation of cytoskeleton tension. J. Cell. Mol. Med. 21(6), 1073–1084 (2017). • Demonstrates the potential beneficial role of utilizing softer 3D culture substrates, such as hydrogels, for improving the retention/expression of pluripotent stem-like genes in MSC populations.
    • 27. Chaicharoenaudomrung N, Kunhorm P, Noisa P. Three-dimensional cell culture systems as an in vitro platform for cancer and stem cell modeling. World J. Stem Cells 11(12), 1065–1083 (2019).
    • 28. Panchalingam KM, Jung S, Rosenberg L, Behie LA. Bioprocessing strategies for the large-scale production of human mesenchymal stem cells: a review. Stem Cell Res. Ther. 6, 225 (2015).
    • 29. Kaminska A, Wedzinska A, Kot M, Sarnowska A. Effect of long-term 3D spheroid culture on WJ-MSC. Cells 10(4), 719 (2021).
    • 30. Tibbitt MW, Anseth KS. Hydrogels as extracellular matrix mimics for 3D cell culture. Biotechnol. Bioeng. 103(4), 655–663 (2009).
    • 31. Caliari SR, Burdick JA. A practical guide to hydrogels for cell culture. Nat. Methods 13(5), 405–414 (2016). • Excellent review describing the fabrication, utilization and benefits of 3D hydrogels for use in cell culture and cell therapies.
    • 32. Tsou YH, Khoneisser J, Huang PC, Xu X. Hydrogel as a bioactive material to regulate stem cell fate. Bioact. Mater. 1(1), 39–55 (2016).
    • 33. Moshayedi P, Nih LR, Llorente IL et al. Systematic optimization of an engineered hydrogel allows for selective control of human neural stem cell survival and differentiation after transplantation in the stroke brain. Biomaterials 105, 145–155 (2016).
    • 34. Chen J, Chin A, Almarza AJ, Taboas JM. Hydrogel to guide chondrogenesis versus osteogenesis of mesenchymal stem cells for fabrication of cartilaginous tissues. Biomed. Mater. 15(4), 045006 (2020).
    • 35. Zigon-Branc S, Markovic M, Van Hoorick J et al. Impact of hydrogel stiffness on differentiation of human adipose-derived stem cell microspheroids. Tissue Eng. A 25(19–20), 1369–1380 (2019).
    • 36. Bonab MM, Alimoghaddam K, Talebian F, Ghaffari SH, Ghavamzadeh A, Nikbin B. Aging of mesenchymal stem cell in vitro. BMC Cell Biol. 7, 14 (2006).
    • 37. Chowdhury F, Li Y, Poh YC, Yokohama-Tamaki T, Wang N, Tanaka TS. Soft substrates promote homogeneous self-renewal of embryonic stem cells via downregulating cell-matrix tractions. PLOS ONE 5(12), e15655 (2010).
    • 38. Gerardo H, Lima A, Carvalho J et al. Soft culture substrates favor stem-like cellular phenotype and facilitate reprogramming of human mesenchymal stem/stromal cells (hMSCs) through mechanotransduction. Sci. Rep. 9(1), 9086 (2019).
    • 39. Wei W, Ma Y, Yao X et al. Advanced hydrogels for the repair of cartilage defects and regeneration. Bioact. Mater. 6(4), 998–1011 (2021).
    • 40. Bai R, Tian L, Li Y et al. Combining ECM hydrogels of cardiac bioactivity with stem cells of high cardiomyogenic potential for myocardial repair. Stem Cells Int. 2019, 6708435 (2019).
    • 41. Wang H, Shi J, Wang Y et al. Promotion of cardiac differentiation of brown adipose derived stem cells by chitosan hydrogel for repair after myocardial infarction. Biomaterials 35(13), 3986–3998 (2014).
    • 42. Distler T, Lauria I, Detsch R et al. Neuronal differentiation from induced pluripotent stem cell-derived neurospheres by the application of oxidized alginate–gelatin–laminin hydrogels. Biomedicines 9(3), 261 (2021).
    • 43. Burnsed OA, Schwartz Z, Marchand KO, Hyzy SL, Olivares-Navarrete R, Boyan BD. Hydrogels derived from cartilage matrices promote induction of human mesenchymal stem cell chondrogenic differentiation. Acta Biomater. 43, 139–149 (2016).
    • 44. Han WM, Mohiuddin M, Anderson SE, Garcia AJ, Jang YC. Co-delivery of Wnt7a and muscle stem cells using synthetic bioadhesive hydrogel enhances murine muscle regeneration and cell migration during engraftment. Acta Biomater. 94, 243–252 (2019).
    • 45. Dimmeler S, Leri A. Aging and disease as modifiers of efficacy of cell therapy. Circ. Res. 102(11), 1319–1330 (2008).
    • 46. Schimke MM, Marozin S, Lepperdinger G. Patient-specific age: the other side of the coin in advanced mesenchymal stem cell therapy. Front. Physiol. 6, 362 (2015).
    • 47. Rodier F, Campisi J. Four faces of cellular senescence. J. Cell Biol. 192(4), 547–556 (2011).
    • 48. Van Deursen JM. The role of senescent cells in ageing. Nature 509(7501), 439–446 (2014).
    • 49. Wagner W, Horn P, Castoldi M et al. Replicative senescence of mesenchymal stem cells: a continuous and organized process. PLOS ONE 3(5), e2213 (2008).
    • 50. Acosta JC, Banito A, Wuestefeld T et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat. Cell Biol. 15(8), 978–990 (2013).
    • 51. Turinetto V, Vitale E, Giachino C. Senescence in human mesenchymal stem cells: functional changes and implications in stem cell-based therapy. Int. J. Mol. Sci. 17(7), 1164 (2016).
    • 52. Ratushnyy A, Ezdakova M, Buravkova L. Secretome of senescent adipose-derived mesenchymal stem cells negatively regulates angiogenesis. Int. J. Mol. Sci. 21(5), 1802 (2020). • One of many articles highlighting some of the negative effects of the secretome and paracrine signaling of MSC populations that have undergone senescence. The authors show how senescent MSC populations can impair angiogenesis.
    • 53. Liu J, Ding Y, Liu Z, Liang X. Senescence in Mesenchymal Stem Cells: Functional Alterations, Molecular Mechanisms, and Rejuvenation Strategies. Front. Cell. Dev. Biol. 8,258(2020).
    • 54. Turinetto V, Vitale E, Giachino C. Senescence in Human Mesenchymal Stem Cells: Functional Changes and Implications in Stem Cell-Based Therapy. Int. J. Mol. Sci. 17(7), 1164 (2016).
    • 55. Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu. Rev. Pathol. 5,99–118 (2010).